1
|
Ege D, Nawaz Q, Beltrán AM, Boccaccini AR. Effect of Boron-Doped Mesoporous Bioactive Glass Nanoparticles on C2C12 Cell Viability and Differentiation: Potential for Muscle Tissue Application. ACS Biomater Sci Eng 2022; 8:5273-5283. [PMID: 36379050 PMCID: PMC9748944 DOI: 10.1021/acsbiomaterials.2c00876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2022]
Abstract
Mesoporous bioactive glasses (MBGs) exhibit a high surface area and a highly ordered mesoporous structure. MBGs have potential for both hard and soft tissue engineering applications. MBGs may be doped with biologically active ions to tailor their biological activity. Boron is being widely studied as a dopant of bioactive glasses. Recently, research has demonstrated the potential of boron-containing bioactive glasses for muscle regeneration. In this study, boron-containing MBGs, 10B-MBG and 18B-MBG nanoparticles, were produced by a microemulsion-assisted sol-gel approach for potential muscle regeneration applications. First, X-ray diffraction (XRD), Fourier transform infrared (FTIR), and energy-dispersive X-ray spectroscopy (EDX) analyses were conducted to study the chemical structure and composition of the nanoparticles. To examine the nanoparticle morphology, scanning electron microscopy (SEM) and transmission electron microscopy (TEM) images were analyzed. Both SEM images and particle size distribution determined by dynamic light scattering (DLS) indicated a decrease of the average particle size after boron doping. TEM images indicated a slit-shaped mesoporous structure of nanoparticles for all compositions. The ζ potential was measured, and a negative surface charge was found for all study groups due to the presence of silanol groups. Cytocompatibility and fluorescence microscopy studies were also carried out. The results indicated that low concentrations (0.1 and 1 mg mL-1) of all MBG nanoparticles led to high viability of C2C12 cells. Fluorescence microscopy images indicated that at lower nanoparticle concentrations (0.1 and 1 mg mL-1), C2C12 cells appeared to differentiate into myotubes, which was indicated by a spindle-shaped morphology. For 10 mg mL-1 concentration of nanoparticles, C2C12 cells had a lower aspect ratio (estimated qualitatively by inspection of the images), which implied a lower degree of differentiation. Boron-doped MBG nanoparticles in reduced concentrations are suitable to induce differentiation of C2C12 cells into myotubes, indicating their potential for applications in muscle tissue repair.
Collapse
Affiliation(s)
- Duygu Ege
- Institute
of Biomedical Engineering, Boğaziçi
University, Rasathane Street, Kandilli, İstanbul34684, Turkey
- Department
of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, 91058Erlangen, Germany
| | - Qaisar Nawaz
- Department
of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, 91058Erlangen, Germany
| | - Ana M. Beltrán
- Departamento
de Ingeniería y Ciencia de los Materiales y del Transporte, Escuela Politécnica Superior, Universidad de
Sevilla, 41011Seville, Spain
| | - Aldo R. Boccaccini
- Department
of Materials Science and Engineering, Institute of Biomaterials, University of Erlangen-Nuremberg, 91058Erlangen, Germany
| |
Collapse
|
2
|
MacDonald A, Gross A, Jones B, Dhar M. Muscle Regeneration of the Tongue: A review of current clinical and regenerative research strategies. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:1022-1034. [PMID: 34693743 DOI: 10.1089/ten.teb.2021.0133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Various abnormalities of the tongue, including cancers, commonly require surgical removal to sequester growth and metastasis. However, even minor resections can affect functional outcomes such as speech and swallowing, thereby reducing quality of life. Surgical resections alone create volumetric muscle loss whereby muscle tissue cannot self-regenerate within the tongue. In these cases, the tongue is reconstructed typically in the form of autologous skin flaps. However, flap reconstruction has many limitations and unfortunately is the primary option for oral and reconstructive surgeons to treat tongue defects. The alternative, but yet undeveloped strategy for tongue reconstruction is regenerative medicine, which widely focuses on building new organs with stem cells. Regenerative medicine has successfully treated many tissues, but research has inadequately addressed the tongue as a vital organ in need of tissue engineering. In this review, we address the current standard for tongue reconstruction, the cellular mechanisms of muscle cell development, and the stem cell studies that have attempted muscle engineering within the tongue. Until now, no review has focused on engineering the tongue with regenerative medicine, which could guide innovative strategies for tongue reconstruction.
Collapse
Affiliation(s)
- Amber MacDonald
- The University of Tennessee Knoxville College of Veterinary Medicine, 70737, Large Animal Clinical Sciences, 2407 River Drive, Knoxville, Tennessee, United States, 37996-4539;
| | - Andrew Gross
- The University of Tennessee Medical Center, 21823, Knoxville, Tennessee, United States;
| | - Brady Jones
- The University of Tennessee Medical Center, 21823, Knoxville, Tennessee, United States;
| | - Madhu Dhar
- University of Tennessee Knoxville College of Veterinary Medicine, 70737, Large Animal Clinical Sciences, College of Veterinary Medicine, 2407 River Drive, Knoxville, Tennessee, United States, 37996.,University of Tennessee;
| |
Collapse
|
3
|
Yang CD, Chuang SC, Cheng TL, Lee MJ, Chen HT, Lin SY, Huang HT, Ho CJ, Lin YS, Kang L, Ho ML, Chang JK, Chen CH. An Intermediate Concentration of Calcium with Antioxidant Supplement in Culture Medium Enhances Proliferation and Decreases the Aging of Bone Marrow Mesenchymal Stem Cells. Int J Mol Sci 2021; 22:ijms22042095. [PMID: 33672524 PMCID: PMC7923799 DOI: 10.3390/ijms22042095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/29/2021] [Accepted: 02/15/2021] [Indexed: 12/26/2022] Open
Abstract
Human bone marrow stem cells (HBMSCs) are isolated from the bone marrow. Stem cells can self-renew and differentiate into various types of cells. They are able to regenerate kinds of tissue that are potentially used for tissue engineering. To maintain and expand these cells under culture conditions is difficult—they are easily triggered for differentiation or death. In this study, we describe a new culture formula to culture isolated HBMSCs. This new formula was modified from NCDB 153, a medium with low calcium, supplied with 5% FBS, extra growth factor added to it, and supplemented with N-acetyl-L-cysteine and L-ascorbic acid-2-phosphate to maintain the cells in a steady stage. The cells retain these characteristics as primarily isolated HBMSCs. Moreover, our new formula keeps HBMSCs with high proliferation rate and multiple linage differentiation ability, such as osteoblastogenesis, chondrogenesis, and adipogenesis. It also retains HBMSCs with stable chromosome, DNA, telomere length, and telomerase activity, even after long-term culture. Senescence can be minimized under this new formulation and carcinogenesis of stem cells can also be prevented. These modifications greatly enhance the survival rate, growth rate, and basal characteristics of isolated HBMSCs, which will be very helpful in stem cell research.
Collapse
Affiliation(s)
- Chung-Da Yang
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912301, Taiwan;
| | - Shu-Chun Chuang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.C.); (T.-L.C.); (S.-Y.L.); (H.-T.H.); (C.-J.H.); (Y.-S.L.); (M.-L.H.)
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Tsung-Lin Cheng
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.C.); (T.-L.C.); (S.-Y.L.); (H.-T.H.); (C.-J.H.); (Y.-S.L.); (M.-L.H.)
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Mon-Juan Lee
- Department of Bioscience Technology, Chang Jung Christian University, Tainan 71101, Taiwan;
- Innovative Research Center of Medicine, Chang Jung Christian University, Tainan 71101, Taiwan
| | - Hui-Ting Chen
- Faculty of Pharmacy, School of Pharmaceutical Sciences, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Fragrance and Cosmetic Science, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Sung-Yen Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.C.); (T.-L.C.); (S.-Y.L.); (H.-T.H.); (C.-J.H.); (Y.-S.L.); (M.-L.H.)
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Hsuan-Ti Huang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.C.); (T.-L.C.); (S.-Y.L.); (H.-T.H.); (C.-J.H.); (Y.-S.L.); (M.-L.H.)
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Cheng-Jung Ho
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.C.); (T.-L.C.); (S.-Y.L.); (H.-T.H.); (C.-J.H.); (Y.-S.L.); (M.-L.H.)
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Yi-Shan Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.C.); (T.-L.C.); (S.-Y.L.); (H.-T.H.); (C.-J.H.); (Y.-S.L.); (M.-L.H.)
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Lin Kang
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
| | - Mei-Ling Ho
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.C.); (T.-L.C.); (S.-Y.L.); (H.-T.H.); (C.-J.H.); (Y.-S.L.); (M.-L.H.)
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Je-Ken Chang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.C.); (T.-L.C.); (S.-Y.L.); (H.-T.H.); (C.-J.H.); (Y.-S.L.); (M.-L.H.)
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Correspondence: (J.-K.C.); (C.-H.C.); Tel.: +886-7-3209-209 (C.-H.C.)
| | - Chung-Hwan Chen
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (S.-C.C.); (T.-L.C.); (S.-Y.L.); (H.-T.H.); (C.-J.H.); (Y.-S.L.); (M.-L.H.)
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Departments of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 80420, Taiwan
- Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Correspondence: (J.-K.C.); (C.-H.C.); Tel.: +886-7-3209-209 (C.-H.C.)
| |
Collapse
|
4
|
Distler T, Solisito AA, Schneidereit D, Friedrich O, Detsch R, Boccaccini AR. 3D printed oxidized alginate-gelatin bioink provides guidance for C2C12 muscle precursor cell orientation and differentiation via shear stress during bioprinting. Biofabrication 2020; 12:045005. [PMID: 32485696 DOI: 10.1088/1758-5090/ab98e4] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Biofabrication can be a tool to three-dimensionally (3D) print muscle cells embedded inside hydrogel biomaterials, ultimately aiming to mimic the complexity of the native muscle tissue and to create in-vitro muscle analogues for advanced repair therapies and drug testing. However, to 3D print muscle analogues of high cell alignment and synchronous contraction, the effect of biofabrication process parameters on myoblast growth has to be understood. A suitable biomaterial matrix is required to provide 3D printability as well as matrix degradation to create space for cell proliferation, matrix remodelling capacity, and cell differentiation. We demonstrate that by the proper selection of nozzle size and extrusion pressure, the shear stress during extrusion-bioprinting of mouse myoblast cells (C2C12) can achieve cell orientation when using oxidized alginate-gelatin (ADA-GEL) hydrogel bionk. The cells grow in the direction of printing, migrate to the hydrogel surface over time, and differentiate into ordered myotube segments in areas of high cell density. Together, our results show that ADA-GEL hydrogel can be a simple and cost-efficient biodegradable bioink that allows the successful 3D bioprinting and cultivation of C2C12 cells in-vitro to study muscle engineering.
Collapse
Affiliation(s)
- Thomas Distler
- Department of Materials Science and Engineering, Institute of Biomaterials, Erlangen 91058, Germany. These authors contributed equally to this work
| | | | | | | | | | | |
Collapse
|
5
|
Chiu CH, Chang TH, Chang SS, Chang GJ, Chen ACY, Cheng CY, Chen SC, Fu JF, Wen CJ, Chan YS. Application of Bone Marrow-Derived Mesenchymal Stem Cells for Muscle Healing After Contusion Injury in Mice. Am J Sports Med 2020; 48:1226-1235. [PMID: 32134689 DOI: 10.1177/0363546520905853] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Skeletal muscle injuries are very common in sports medicine. Conventional therapies have limited clinical efficacy. New treatment methods should be developed to allow athletes to return to play with better function. PURPOSE To evaluate the in vitro differentiation potential of bone marrow-derived mesenchymal stem cells and the in vivo histologic and physiologic effects of mesenchymal stem cell therapy on muscle healing after contusion injury. STUDY DESIGN Controlled laboratory study. METHODS Bone marrow cells were flushed from both femurs of 5-week-old C57BL/6 mice to establish immortalized mesenchymal stem cell lines. A total of 36 mice aged 8 to 10 weeks were used to develop a muscle contusion model and were divided into 6 groups (6 mice/group) on the basis of the different dosages of IM2 cells to be injected (0, 1.25 × 105, and 2.5 × 105 cells with/without F-127 in 100 μL of phosphate-buffered saline). Histological analysis of muscle regeneration was performed, and the fast-twitch and tetanus strength of the muscle contractions was measured 28 days after muscle contusion injury, after injections of different doses of mesenchymal stem cells with or without the F-127 scaffold beginning 14 days after contusion injury. RESULTS The mesenchymal stem cell-treated muscles exhibited numerous regenerating myofibers. All the groups treated with mesenchymal stem cells (1.25 × 105 cells, 2.5 × 105 cells, 1.25 × 105 cells plus F-127, and 2.5 × 105 cells plus F-127) exhibited a significantly higher number of regenerating myofibers (mean ± SD: 111.6 ± 14.77, 133.4 ± 21.44, 221.89 ± 32.65, and 241.5 ± 25.95, respectively) as compared with the control group and the control with F-127 (69 ± 18.79 and 63.2 ± 18.98). The physiologic evaluation of fast-twitch and tetanus strength did not reveal differences between the age-matched uninjured group and the groups treated with various doses of mesenchymal stem cells 28 days after contusion. Significant differences were found between the control group and the groups treated with various doses of mesenchymal stem cells after muscle contusion. CONCLUSION Mesenchymal stem cell therapy increased the number of regenerating myofibers and improved fast-twitch and tetanus muscle strength in a mouse model of muscle contusion. However, the rapid decay of transplanted mesenchymal stem cells suggests a paracrine effect of this action. Treatment with mesenchymal stem cells at various doses combined with the F-127 scaffold is a potential therapy for a muscle contusion. CLINICAL RELEVANCE Mesenchymal stem cell therapy has an effect on sports medicine because of its effects on myofiber regeneration and muscle strength after contusion injury.
Collapse
Affiliation(s)
- Chih-Hao Chiu
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan.,Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou
| | - Tsan-Hsuan Chang
- Department of General Medicine, Tri-service General Hospital, Taipei
| | - Shih-Sheng Chang
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan.,Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou
| | - Gwo-Jyh Chang
- Graduate Institute of Clinical and Medicinal Sciences, College of Medicine, Chang Gung University, Taoyuan
| | - Alvin Chao-Yu Chen
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou.,Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Linkou
| | - Chun-Ying Cheng
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou.,Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Linkou
| | - Su-Ching Chen
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou.,Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Linkou
| | - Jen-Fen Fu
- Department of Medical Research, Chang Gung Memorial Hospital, Linkou.,Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan
| | - Chih-Jen Wen
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Linkou.,College of Medicine, Chang Gung University, Taoyuan
| | - Yi-Sheng Chan
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou.,Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Linkou
| |
Collapse
|
6
|
Galli F, Bragg L, Meggiolaro L, Rossi M, Caffarini M, Naz N, Santoleri S, Cossu G. Gene and Cell Therapy for Muscular Dystrophies: Are We Getting There? Hum Gene Ther 2019; 29:1098-1105. [PMID: 30132372 PMCID: PMC6211823 DOI: 10.1089/hum.2018.151] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
In the last few years, significant advances have occurred in the preclinical and clinical work toward gene and cell therapy for muscular dystrophy. At the time of this writing, several trials are ongoing and more are expected to start. It is thus a time of expectation, even though many hurdles remain and it is unclear whether they will be overcome with current strategies or if further improvements will be necessary.
Collapse
Affiliation(s)
- Francesco Galli
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester , Manchester, United Kingdom
| | - Laricia Bragg
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester , Manchester, United Kingdom
| | - Linda Meggiolaro
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester , Manchester, United Kingdom
| | - Maira Rossi
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester , Manchester, United Kingdom
| | - Miriam Caffarini
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester , Manchester, United Kingdom
| | - Naila Naz
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester , Manchester, United Kingdom
| | - Sabrina Santoleri
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester , Manchester, United Kingdom
| | - Giulio Cossu
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester , Manchester, United Kingdom
| |
Collapse
|
7
|
Dai M, Yu M, Zhang Y, Tian W. Exosome-Like Vesicles Derived from Adipose Tissue Provide Biochemical Cues for Adipose Tissue Regeneration. Tissue Eng Part A 2017; 23:1221-1230. [PMID: 28457190 DOI: 10.1089/ten.tea.2017.0045] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
There is an emerging need for soft tissue replacements in the field of reconstructive surgery for the treatment of congenital deformities, posttraumatic repair, and cancer rehabilitation. Previous studies have shown that the bioactive adipose tissue extract can induce adipogenesis without additional stem cells or growth factors. In this study, we innovatively investigated whether exosome-like vesicles derived from adipose tissue (ELV-AT) could direct stem cell differentiation and trigger adipose tissue regeneration. In vitro, ELV-AT can induce adipogenesis of adipose-derived stem cells and promote proliferation, migration, and angiogenic potential of the aorta endothelial cells. In vivo, ELV-AT were transplanted to a chamber on the back of nude mice and neoadipose tissues were formed. Our findings indicated that ELV-AT could be used as a cell-free therapeutic approach for adipose tissue regeneration.
Collapse
Affiliation(s)
- Minjia Dai
- 1 State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University , Chengdu, China .,2 National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University , Chengdu, China .,3 Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University , Chengdu, China
| | - Mei Yu
- 1 State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University , Chengdu, China .,2 National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University , Chengdu, China
| | - Yan Zhang
- 1 State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University , Chengdu, China .,2 National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University , Chengdu, China .,3 Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University , Chengdu, China
| | - Weidong Tian
- 1 State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University , Chengdu, China .,2 National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University , Chengdu, China .,3 Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University , Chengdu, China
| |
Collapse
|
8
|
Bana N, Sanooghi D, Soleimani M, Hayati Roodbari N, Alavi Moghaddam S, Joghataei MT, Sayahpour FA, Faghihi F. A Comparative Study to Evaluate Myogenic Differentiation Potential of Human Chorion versus Umbilical Cord Blood-derived Mesenchymal Stem Cells. Tissue Cell 2017; 49:495-502. [PMID: 28601197 DOI: 10.1016/j.tice.2017.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 05/01/2017] [Accepted: 05/10/2017] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Musculodegenerative diseases threaten the life of many patients in the world. Since drug administration is not efficient in regeneration of damaged tissues, stem cell therapy is considered as a good strategy to restore the lost cells. Since the efficiency of myogenic differentiation potential of human Chorion- derived Mesenchymal Stem Cells (C-MSCs) has not been addressed so far; we set out to evaluate myogenic differentiation property of these cells in comparison with Umbilical Cord Blood- derived Mesenchymal Stem Cells (UCB-MSCs) in the presence of 5-azacytidine. MATERIALS & METHODS To do that, neonate placenta Umbilical Cord Blood were transferred to the lab. After characterization of the isolated cells using flowcytometry and multilineage differentiation capacity, the obtained Mesenchymal Stem Cells were cultured in DMEM/F12 supplemented with 2% FBS and 10μM of 5-azacytidine to induce myogenic differentiation. Real-time PCR and immunocytochemistry were used to assess the myogenic properties of the cells. RESULTS Our data showed that C-MSCs and UCB-MSCs were spindle shape in morphology. They were positive for CD90, CD73 and CD44 antigens, and negative for hematopoietic markers. They also differentiated into osteoblast and adipoblast lineages. Real-time PCR results showed that the cells could express MyoD, desmin and α-MHC at the end of the first week (P<0.05). No significant upregulation was detected in the expression of GATA-4 in both groups. Immunocytochemical staining revealed the expression of Desmin, cTnT and α-MHC. CONCLUSIONS Results showed that these cells are potent to differentiate into myoblast- like cells. An upregulation in the expression of some myogenic markers (desmin, α- MHC) was observed in C-MSCs in comparison with UCB-MSCs.
Collapse
Affiliation(s)
- Nikoo Bana
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Davood Sanooghi
- Department of Genetics, Faculty of Biological Sciences, Shahid Beheshti University, Tehran, Iran
| | - Mansoureh Soleimani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nasim Hayati Roodbari
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Mohammad Taghi Joghataei
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Forough Azam Sayahpour
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Faezeh Faghihi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Helal MAM, Shaheen NEM, Abu Zahra FA. Immunomodulatory capacity of the local mesenchymal stem cells transplantation after severe skeletal muscle injury in female rats. Immunopharmacol Immunotoxicol 2016; 38:414-422. [PMID: 27560658 DOI: 10.1080/08923973.2016.1222617] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
CONTEXT Cell therapy technique with stem cells is a very attractive strategy for the treatment of muscle disorders. OBJECTIVE The objective of this study was to investigate the mechanism of local transplantation of mesenchymal stem cells (MSCs) which could contribute to skeletal muscle healing. MATERIALS AND METHODS Female rats were divided into three equal groups as the following: group 1, the negative control group (untreated group), group 2, sham-treated group, rats with muscle injuries involving volumetric muscle loss (VML) of adductor brevis muscle and injected locally with phosphate-buffered saline (PBS) 0.5 ml without stem cells after 7 d of muscle injury, group 3, treated group, rats with VML and injected locally (intramuscular) with 1.5 × 106 bone marrow MSCs suspended in PBS 0.5 ml (1) after 7 d of muscle tissue injury. All animals were sacrificed after 4 weeks of stem cell transplantation. RESULTS In vitro culture the morphology of MSCs reached confluence and appeared as long spindle in shape on 9-14 d. Most of the cells did not express the hematopoietic cell marker, CD34 and CD45 but expressed MSCs marker CD44, CD90 and CD105. The remarkable increase of proliferating cell nuclear antigen positive nucleus was recorded in MSCs group as compared to PBS group. After 28 d of injection, administration of only PBS into the site of muscle injury caused up-regulation in the levels of interleukins IL-1β, IL-6, tumor necrosis factor alpha (TNF-α), transforming growth factor beta (TGF-β1), interferon alpha (IFN-α) and down-regulate the level of IL-10 in muscular tissue comparing to the untreated control. Bone marrow MSCs + PBS injected at the site of muscle injury significantly down-regulate the inflammatory cytokines levels IL-1β and IL-6 and TNF-α, TGF-β1 and IFN-α and up-regulate the level of IL-10. Collagen concentrations in the injured skeletal muscle estimated by enzyme-linked immuno sorbent assay and stained with Masson trichrome stain were increased with PBS group and decreased after transplantation of bone marrow MSCs in the site of injury. Muscle sections stained with H&E showed a higher number of centronucleated regenerating myofibers in the stem-cell-treated group than in the (PBS) and untreated control group. Microvasculature of skeletal muscle was decreased as demonstrated by immunostaining technique for CD34 in PBS group from untreated control. The MSCs group showed angiogenesis and marked increase of skeletal muscle microvasculature than PBS group. CONCLUSION MSCs can modify the local immunological responses and improve muscle regeneration by suppressing of inflammatory cytokines, activating of the anti-inflammatory cytokine, restoration of muscle fibers and angiogenesis. By means of increase in TGF-β production in response to muscle injury prevent the repair of injured fibers and increase connective tissue production (collagen fibers), thus propagating skeletal muscle weakness and fibrosis whereas MSCs + PBS injected at the site of muscle injury significantly down-regulate (TGF-β1) and hence the level of collagen (fibrosis or scar areas). MSCs are able to block the fibrotic signaling cascade by declining TGF-β1 and scar areas in the injured muscle.
Collapse
Affiliation(s)
- Mona A M Helal
- a Department of Zoology, Faculty of Women for Arts, Science & Education , Ain Shams University , Cairo , Egypt
| | - Noura E M Shaheen
- a Department of Zoology, Faculty of Women for Arts, Science & Education , Ain Shams University , Cairo , Egypt
| | - Fatma A Abu Zahra
- b Molecular Biology and Tissue Culture , Medical Research Center, Ain Shams University , Cairo , Egypt
| |
Collapse
|
10
|
Developmental Biology and Regenerative Medicine: Addressing the Vexing Problem of Persistent Muscle Atrophy in the Chronically Torn Human Rotator Cuff. Phys Ther 2016; 96:722-33. [PMID: 26847008 PMCID: PMC4858662 DOI: 10.2522/ptj.20150029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 01/24/2016] [Indexed: 12/18/2022]
Abstract
Persistent muscle atrophy in the chronically torn rotator cuff is a significant obstacle for treatment and recovery. Large atrophic changes are predictive of poor surgical and nonsurgical outcomes and frequently fail to resolve even following functional restoration of loading and rehabilitation. New insights into the processes of muscle atrophy and recovery gained through studies in developmental biology combined with the novel tools and strategies emerging in regenerative medicine provide new avenues to combat the vexing problem of muscle atrophy in the rotator cuff. Moving these treatment strategies forward likely will involve the combination of surgery, biologic/cellular agents, and physical interventions, as increasing experimental evidence points to the beneficial interaction between biologic therapies and physiologic stresses. Thus, the physical therapy profession is poised to play a significant role in defining the success of these combinatorial therapies. This perspective article will provide an overview of the developmental biology and regenerative medicine strategies currently under investigation to combat muscle atrophy and how they may integrate into the current and future practice of physical therapy.
Collapse
|
11
|
Wang X, Shen QW, Wang J, Zhang Z, Feng F, Chen T, Zhang Y, Wei H, Li Z, Wang X, Wang Y. KLF7 Regulates Satellite Cell Quiescence in Response to Extracellular Signaling. Stem Cells 2016; 34:1310-20. [PMID: 26930448 DOI: 10.1002/stem.2346] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 11/12/2015] [Indexed: 11/11/2022]
Abstract
Retaining muscle stem satellite cell (SC) quiescence is important for the maintenance of stem cell population and tissue regeneration. Accumulating evidence supports the model where key extracellular signals play crucial roles in maintaining SC quiescence or activation, however, the intracellular mechanisms that mediate niche signals to control SC behavior are not fully understood. Here, we reported that KLF7 functioned as a key mediator involved in low-level TGF-β signaling and canonical Notch signaling-induced SC quiescence and myoblast arrest. The data obtained showed that KLF7 was upregulated in quiescent SCs and nonproliferating myoblasts. Silence of KLF7 promoted SCs activation and myoblasts proliferation, but overexpression of KLF7 induced myogenic cell arrest. Notably, the expression of KLF7 was regulated by TGF-β and Notch3 signaling. Knockdown of KLF7 diminished low-level TGF-β and canonical Notch signaling-induced SC quiescence. Investigation into the mechanism revealed that KLF7 regulation of SC function was dependent on p21 and acetylation of Lys227 and/or 231 in the DNA binding domain of KLF7. Our study provides new insights into the regulatory network of muscle stem cell quiescence. Stem Cells 2016;34:1310-1320.
Collapse
Affiliation(s)
- Xiaobin Wang
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Qingwu W Shen
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, People's Republic of China.,College of Food Science and Technology, Hunan Agricultural University, Changsha, Hunan, People's Republic of China
| | - Jie Wang
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Zhiguo Zhang
- College of Food Science and Engineering, Qilu University of Technology, Jinan, Shandong, People's Republic of China
| | - Fu Feng
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Ting Chen
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Yanyan Zhang
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Huan Wei
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Zhongwen Li
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Xinxia Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Yizhen Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
12
|
Joanisse S, Parise G. Cytokine Mediated Control of Muscle Stem Cell Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 900:27-44. [DOI: 10.1007/978-3-319-27511-6_2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
13
|
Choi JS, Yoon HI, Lee KS, Choi YC, Yang SH, Kim IS, Cho YW. Exosomes from differentiating human skeletal muscle cells trigger myogenesis of stem cells and provide biochemical cues for skeletal muscle regeneration. J Control Release 2015; 222:107-15. [PMID: 26699421 DOI: 10.1016/j.jconrel.2015.12.018] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 12/03/2015] [Accepted: 12/12/2015] [Indexed: 12/19/2022]
Abstract
Exosomes released from skeletal muscle cells play important roles in myogenesis and muscle development via the transfer of specific signal molecules. In this study, we investigated whether exosomes secreted during myotube differentiation from human skeletal myoblasts (HSkM) could induce a cellular response from human adipose-derived stem cells (HASCs) and enhance muscle regeneration in a muscle laceration mouse model. The exosomes contained various signal molecules including myogenic growth factors related to muscle development, such as insulin-like growth factors (IGFs), hepatocyte growth factor (HGF), fibroblast growth factor-2 (FGF2), and platelet-derived growth factor-AA (PDGF-AA). Interestingly, exosome-treated HASCs fused with neighboring cells at early time points and exhibited a myotube-like phenotype with increased expression of myogenic proteins (myosin heavy chain and desmin). On day 21, mRNAs of terminal myogenic genes were also up-regulated in exosome-treated HASCs. Moreover, in vivo studies demonstrated that exosomes from differentiating HSkM reduced the fibrotic area and increased the number of regenerated myofibers in the injury site, resulting in significant improvement of skeletal muscle regeneration. Our findings suggest that exosomes act as a biochemical cue directing stem cell differentiation and provide a cell-free therapeutic approach for muscle regeneration.
Collapse
Affiliation(s)
- Ji Suk Choi
- Department of Chemical Engineering, Hanyang University, Ansan, Gyeonggi-do 426-791, Republic of Korea
| | - Hwa In Yoon
- Department of Chemical Engineering, Hanyang University, Ansan, Gyeonggi-do 426-791, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 136-791, Republic of Korea
| | - Kyoung Soo Lee
- Department of Chemical Engineering, Hanyang University, Ansan, Gyeonggi-do 426-791, Republic of Korea
| | - Young Chan Choi
- Department of Chemical Engineering, Hanyang University, Ansan, Gyeonggi-do 426-791, Republic of Korea
| | - Seong Hyun Yang
- Department of Chemical Engineering, Hanyang University, Ansan, Gyeonggi-do 426-791, Republic of Korea
| | - In-San Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 136-791, Republic of Korea; KU-KIST School, Korea University, 1 Anam-dong, Seongbuk-gu, Seoul 136-701, Republic of Korea
| | - Yong Woo Cho
- Department of Chemical Engineering, Hanyang University, Ansan, Gyeonggi-do 426-791, Republic of Korea.
| |
Collapse
|
14
|
Zhang Y, Zhu Y, Li Y, Cao J, Zhang H, Chen M, Wang L, Zhang C. Long-term engraftment of myogenic progenitors from adipose-derived stem cells and muscle regeneration in dystrophic mice. Hum Mol Genet 2015; 24:6029-40. [DOI: 10.1093/hmg/ddv316] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 07/31/2015] [Indexed: 12/12/2022] Open
|
15
|
Abstract
Regenerative capacity of skeletal muscles resides in satellite cells, a self-renewing population of muscle cells. Several studies are investigating epigenetic mechanisms that control myogenic proliferation and differentiation to find new approaches that could boost regeneration of endogenous myogenic progenitor populations. In recent years, a lot of effort has been applied to purify, expand and manipulate adult stem cells from muscle tissue. However, this population of endogenous myogenic progenitors in adults is limited and their access is difficult and invasive. Therefore, other sources of stem cells with potential to regenerate muscles need to be examined. An excellent candidate could be a population of adult stromal cells within fat characterized by mesenchymal properties, which have been termed adipose-derived stem cells (ASCs). These progenitor adult stem cells have been successfully differentiated in vitro to osteogenic, chondrogenic, neurogenic and myogenic lineages. Autologous ASCs are multipotent and can be harvested with low morbidity; thus, they hold promise for a range of therapeutic applications. This review will summarize the use of ASCs in muscle regenerative approaches.
Collapse
Affiliation(s)
- Sonia-V Forcales
- Genetics and Epigenetics of Cancer, Institute of Predictive and Personalized Medicine of Cancer Barcelona, Spain
| |
Collapse
|
16
|
Mutlu L, Hufnagel D, Taylor HS. The endometrium as a source of mesenchymal stem cells for regenerative medicine. Biol Reprod 2015; 92:138. [PMID: 25904012 DOI: 10.1095/biolreprod.114.126771] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 04/09/2015] [Indexed: 12/21/2022] Open
Abstract
Stem cell therapies have opened new frontiers in medicine with the possibility of regenerating lost or damaged cells. Embryonic stem cells, induced pluripotent stem cells, hematopoietic stem cells, and mesenchymal stem cells have been used to derive mature cell types for tissue regeneration and repair. However, the endometrium has emerged as an attractive, novel source of adult stem cells that are easily accessed and demonstrate remarkable differentiation capacity. In this review, we summarize our current understanding of endometrial stem cells and their therapeutic potential in regenerative medicine.
Collapse
Affiliation(s)
- Levent Mutlu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Demetra Hufnagel
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
17
|
Mesenchymal stromal cells from adipose tissue attached to suture material enhance the closure of enterocutaneous fistulas in a rat model. Cytotherapy 2014; 16:1709-19. [DOI: 10.1016/j.jcyt.2014.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 08/12/2014] [Accepted: 08/15/2014] [Indexed: 02/07/2023]
|
18
|
Alexeev V, Arita M, Donahue A, Bonaldo P, Chu ML, Igoucheva O. Human adipose-derived stem cell transplantation as a potential therapy for collagen VI-related congenital muscular dystrophy. Stem Cell Res Ther 2014; 5:21. [PMID: 24522088 PMCID: PMC4054951 DOI: 10.1186/scrt411] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 12/20/2013] [Indexed: 12/16/2022] Open
Abstract
Introduction Congenital muscular dystrophies (CMD) are a clinically and genetically heterogeneous group of neuromuscular disorders characterized by muscle weakness within the first two years of life. Collagen VI-related muscle disorders have recently emerged as one of the most common types of CMD. COL6 CMD is caused by deficiency and/or dysfunction of extracellular matrix (ECM) protein collagen VI. Currently, there is no specific treatment for this disabling and life-threatening disease. The primary cellular targets for collagen VI CMD therapy are fibroblasts in muscle, tendon and skin, as opposed to muscle cells for other types of muscular dystrophies. However, recent advances in stem cell research have raised the possibility that use of adult stem cells may provide dramatic new therapies for treatment of COL6 CMD. Methods Here, we developed a procedure for isolation of human stem cells from the adipose layer of neonatal skin. The adipose-derived stem cells (ADSC) were examined for expression of ECM and related genes using gene expression array analysis. The therapeutic potential of ADSC was assessed after a single intramuscular transplantation in collagen VI-deficient mice. Results Analysis of primary cultures confirmed that established ADSC represent a morphologically homogenous population with phenotypic and functional features of adult mesenchymal stem cells. A comprehensive gene expression analysis showed that ADSC express a vast array of ECM genes. Importantly, it was observed that ADSC synthesize and secrete all three collagen VI chains, suggesting suitability of ADSC for COL6 CMD treatment. Furthermore, we have found that a single intramuscular transplantation of ADSC into Col6a1−/−Rag1−/− mice under physiological and cardiotoxin-induced injury/regeneration conditions results in efficient engraftment and migration of stem cells within the skeletal muscle. Importantly, we showed that ADSC can survive long-term and continuously secrete the therapeutic collagen VI protein missing in the mutant mice. Conclusions Overall, our findings suggest that stem cell therapy can potentially provide a new avenue for the treatment of COL6 CMD and other muscular disorders and injuries.
Collapse
|
19
|
Ninagawa NT, Isobe E, Hirayama Y, Murakami R, Komatsu K, Nagai M, Kobayashi M, Kawabata Y, Torihashi S. Transplantated mesenchymal stem cells derived from embryonic stem cells promote muscle regeneration and accelerate functional recovery of injured skeletal muscle. Biores Open Access 2013; 2:295-306. [PMID: 23914336 PMCID: PMC3731682 DOI: 10.1089/biores.2013.0012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We previously established that mesenchymal stem cells originating from mouse embryonic stem (ES) cells (E-MSCs) showed markedly higher potential for differentiation into skeletal muscles in vitro than common mesenchymal stem cells (MSCs). Further, the E-MSCs exhibited a low risk for teratoma formation. Here we evaluate the potential of E-MSCs for differentiation into skeletal muscles in vivo and reveal the regeneration and functional recovery of injured muscle by transplantation. E-MSCs were transplanted into the tibialis anterior (TA) muscle 24 h following direct clamping. After transplantation, the myogenic differentiation of E-MSCs, TA muscle regeneration, and re-innervation were morphologically analyzed. In addition, footprints and gaits of each leg under spontaneous walking were measured by CatWalk XT, and motor functions of injured TA muscles were precisely analyzed. Results indicate that >60% of transplanted E-MSCs differentiated into skeletal muscles. The cross-sectional area of the injured TA muscles of E-MSC–transplanted animals increased earlier than that of control animals. E-MSCs also promotes re-innervation of the peripheral nerves of injured muscles. Concerning function of the TA muscles, we reveal that transplantation of E-MSCs promotes the recovery of muscles. This is the first report to demonstrate by analysis of spontaneous walking that transplanted cells can accelerate the functional recovery of injured muscles. Taken together, the results show that E-MSCs have a high potential for differentiation into skeletal muscles in vivo as well as in vitro. The transplantation of E-MSCs facilitated the functional recovery of injured muscles. Therefore, E-MSCs are an efficient cell source in transplantation.
Collapse
Affiliation(s)
- Nana Takenaka Ninagawa
- Department of Rehabilitation Sciences, Graduate School of Medicine, Nagoya University , Nagoya, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Piccioni A, Gaetani E, Neri V, Gatto I, Palladino M, Silver M, Smith RC, Giarretta I, Pola E, Hlatky L, Pola R. Sonic hedgehog therapy in a mouse model of age-associated impairment of skeletal muscle regeneration. J Gerontol A Biol Sci Med Sci 2013; 69:245-52. [PMID: 23781099 DOI: 10.1093/gerona/glt076] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Sonic hedgehog (Shh) is a morphogen regulating muscle development during embryogenesis. We have shown that the Shh pathway is postnatally recapitulated after injury and during regeneration of the adult skeletal muscle and regulates angiogenesis and myogenesis after muscle injury. Here, we demonstrate that in 18-month-old mice, there is a significant impairment of the upregulation of the Shh pathway that physiologically occurs in the young skeletal muscle after injury. Such impairment is even more pronounced in 24-month-old mice. In old animals, intramuscular therapy with a plasmid encoding the human Shh gene increases the regenerative capacities of the injured muscle, in terms of Myf5-positive cells, regenerating myofibers, and fibrosis. At the molecular level, Shh treatment increases the upregulation of the prototypical growth factors, insulin-like growth factor-1 and vascular endothelial growth factor. These data demonstrate that Shh increases regeneration after injury in the muscle of 24-month-old mice and suggest that the manipulation of the Shh pathway may be useful for the treatment of muscular diseases associated with aging.
Collapse
Affiliation(s)
- Andrea Piccioni
- Center of Cancer Systems Biology, CBR4, St. Elizabeth's Medical Center, Tufts University School of Medicine, 736 Cambridge Street, Boston, MA 02135.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Synergistic effects of GDNF and VEGF on lifespan and disease progression in a familial ALS rat model. Mol Ther 2013; 21:1602-10. [PMID: 23712039 DOI: 10.1038/mt.2013.108] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 04/21/2013] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of motor neurons in the brain and spinal cord. We have recently shown that human mesenchymal stem cells (hMSCs) modified to release glial cell line-derived neurotrophic factor (GDNF) decrease disease progression in a rat model of ALS when delivered to skeletal muscle. In the current study, we determined whether or not this effect could be enhanced by delivering GDNF in concert with other trophic factors. hMSC engineered to secrete GDNF (hMSC-GDNF), vascular endothelial growth factor (hMSC-VEGF), insulin-like growth factor-I (hMSC-IGF-I), or brain-derived neurotrophic factor (hMSC-BDNF), were prepared and transplanted bilaterally into three muscle groups. hMSC-GDNF and hMSC-VEGF prolonged survival and slowed the loss of motor function, but hMSC-IGF-I and hMSC-BDNF did not have any effect. We then tested the efficacy of a combined ex vivo delivery of GDNF and VEGF in extending survival and protecting neuromuscular junctions (NMJs) and motor neurons. Interestingly, the combined delivery of these neurotrophic factors showed a strong synergistic effect. These studies further support ex vivo gene therapy approaches for ALS that target skeletal muscle.
Collapse
|
22
|
Olguín HC, Pisconti A. Marking the tempo for myogenesis: Pax7 and the regulation of muscle stem cell fate decisions. J Cell Mol Med 2012; 16:1013-25. [PMID: 21615681 PMCID: PMC4365881 DOI: 10.1111/j.1582-4934.2011.01348.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Post-natal growth and regeneration of skeletal muscle is highly dependent on a population of resident myogenic precursors known as satellite cells. Transcription factors from the Pax gene family, Pax3 and Pax7, are critical for satellite cell biogenesis, survival and potentially self-renewal; however, the underlying molecular mechanisms remain unsolved. This is particularly true in the case of Pax7, which appears to regulate myogenesis at multiple levels. Accordingly, recent data have highlighted the importance of a functional relationship between Pax7 and the MyoD family of muscle regulatory transcription factors during normal muscle formation and disease. Here we will critically review key findings suggesting that Pax7 may play a dual role by promoting resident muscle progenitors to commit to the skeletal muscle lineage while preventing terminal differentiation, thus keeping muscle progenitors poised to differentiate upon environmental cues. In addition, potential regulatory mechanisms for the control of Pax7 activity will be proposed.
Collapse
Affiliation(s)
- Hugo C Olguín
- Departamento Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | | |
Collapse
|
23
|
Zhang Y, Mu Q, Zhou H, Vrijens K, Roussel MF, Jiang G, Yan B. Binding of carbon nanotube to BMP receptor 2 enhances cell differentiation and inhibits apoptosis via regulating bHLH transcription factors. Cell Death Dis 2012; 3:e308. [PMID: 22573038 PMCID: PMC3366082 DOI: 10.1038/cddis.2012.48] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 03/19/2012] [Accepted: 04/02/2012] [Indexed: 12/11/2022]
Abstract
Biomaterials that can drive stem cells to an appropriate differentiation level and decrease apoptosis of transplanted cells are needed in regenerative medicine. Nanomaterials are promising novel materials for such applications. Here we reported that carboxylated multiwalled carbon nanotube (MWCNT 1) promotes myogenic differentiation of mouse myoblast cells and inhibits cell apoptosis under the differentiation conditions by regulating basic helix-loop-helix transcription factors. MWCNT 1 attenuates bone morphogenetic protein receptor (BMPR) signaling activity by binding to BMPR2 and attenuating the phosphorylation of BMPR1. This molecular understanding allowed us to tune stem cell differentiation to various levels by chemical modifications, demonstrating human control of biological activities of nanoparticles and opening an avenue for potential applications of nanomaterials in regenerative medicine.
Collapse
Affiliation(s)
- Y Zhang
- School of Pharmaceutical Sciences, Shandong University, Jinan 250100, China
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Q Mu
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - H Zhou
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - K Vrijens
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - M F Roussel
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - G Jiang
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - B Yan
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China
| |
Collapse
|
24
|
Peçanha R, Bagno LDLES, Ribeiro MB, Robottom Ferreira AB, Moraes MO, Zapata-Sudo G, Kasai-Brunswick TH, Campos-de-Carvalho AC, Goldenberg RCDS, Saar Werneck-de-Castro JP. Adipose-derived stem-cell treatment of skeletal muscle injury. J Bone Joint Surg Am 2012; 94:609-17. [PMID: 22488617 DOI: 10.2106/jbjs.k.00351] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND The aim of the present study was to investigate whether adipose-derived stem cells could contribute to skeletal muscle-healing. METHODS Adipose-derived stem cells of male rats were cultured and injected into the soleus muscles of female rats. Two and four weeks after injections, muscles were tested for tetanic force (50 Hz). Histological analysis was performed to evaluate muscle collagen deposition and the number of centronucleated muscle fibers. In order to track donor cells, chimerism was detected with use of real-time polymerase chain reaction targeting the male sex-determining region Y (SRY) gene. RESULTS Two weeks after cell injection, tetanus strength and the number of centronucleated regenerating myofibers, as well as the number of centronucleated regenerating myofibers, were higher in the treated group than they were in the control group (mean and standard error of the mean, 79.2 ± 5.0% versus 58.3 ± 8.1%, respectively [p < 0.05]; and 145 ± 36 versus 273 ± 18 per 10³ myofibers, respectively [p < 0.05]). However, there were no significant differences at four weeks. Treatment did not decrease collagen deposition. Male gene was not detected in female host tissue at two and four weeks after engraftment by polymerase chain reaction analysis. CONCLUSIONS Adipose-derived stem-cell therapy increased muscle repair and force at two weeks, but not four weeks, after injection, suggesting that adipose-derived stem-cell administration may accelerate muscle repair; however, the rapid disappearance of injected cells suggests a paracrine mechanism of action.
Collapse
Affiliation(s)
- Ramon Peçanha
- Escola de Educação Física e Desportos-CCS, Laboratório de Biologia do Exercício, Departamento de Biociência e Atividade Física, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 540 Ilha do Fundão, Rio de Janeiro, 21941-599, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
The efficiency of in vitro isolation and myogenic differentiation of MSCs derived from adipose connective tissue, bone marrow, and skeletal muscle tissue. In Vitro Cell Dev Biol Anim 2012; 48:203-15. [PMID: 22396125 DOI: 10.1007/s11626-012-9488-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 01/19/2012] [Indexed: 12/19/2022]
Abstract
The objective of the study is to evaluate efficiency of in vitro isolation and myogenic differentiation of mesenchymal stem cells (MSCs) derived from adipose connective tissue (AD-MSCs), bone marrow (BM-MSCs), and skeletal muscle tissue (MC-MSCs). MSCs were isolated from adipose connective tissue, bone marrow, and skeletal muscle tissue of two adult 6-wk-old rats. Cultured MSCs were treated with 5-azacytidine (AZA) to induce myogenic differentiation. Isolated MSCs and differentiated cells were evaluated by immunocytochemistry (ICC), fluorescence-activated cell sorting (FACS), PCR, and RT-PCR. AD-MSCs showed the highest proliferation rate while BM-MSCs had the lowest one. In ICC, isolated MSCs had strong CD90- and CD44-positive expression and negative expression of CD45, CD31, and CD34, while AZA-treated MSCs had strong positive desmin expression. In FACS analysis, AD-MSCs had the highest percentage of CD90- and CD44-positive-expressing cells (99% and 96%) followed by BM-MSCs (97% and 94%) and MC-MSCs (92% and 91%).At 1 wk after incubation with AZA treatment, the peak of myogenin expression reached 93% in differentiated MC-MSCs, 83.3% in BM-MSCs, and 77% in AD-MSCs. MSCs isolated from adipose connective tissue, bone marrow, and skeletal muscle tissue have the same morphology and phenotype, but AD-MSCs were the most easily accessible and had the highest rate of growth on cultivation and the highest percentage of stem cell marker expression. Moreover, although MC-MSCs showed the highest rate of myogenic differentiation potential and expression of myoblast markers, AD-MSCs and BM-MSCs still can be valuable alternatives. The differentiated myoblastic cells could be an available new choice for myoblastic auto-transplantation in regeneration medicine.
Collapse
|
26
|
Arakawa R, Aoki R, Arakawa M, Saito K. Human first-trimester chorionic villi have a myogenic potential. Cell Tissue Res 2012; 348:189-97. [PMID: 22370594 PMCID: PMC3316778 DOI: 10.1007/s00441-012-1340-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 01/18/2012] [Indexed: 12/13/2022]
Abstract
First-trimester chorionic-villi-derived cells (FTCVs) are the earliest fetal material that can be obtained for prenatal diagnosis of fetal disorders such as Duchenne muscular dystrophy (DMD). DMD is a devastating X-linked disorder characterized by the absence of dystrophin at the sarcolemma of muscle fibers. Currently, a limited number of treatment options are available for DMD, although cell therapy is a promising treatment strategy for muscle degeneration in DMD patients. A novel candidate source of cells for this approach is FTCVs taken between the 9th and 11th weeks of gestation. FTCVs might have a higher undifferentiated potential than any other tissue-derived cells because they are the earliest fetal material. We examined the expression of mesenchymal stem cell and pluripotent stem cell markers in FTCVs, in addition to their myogenic potential. FTCVs expressed mesenchymal stem cell markers and Nanog and Sox2 transcription factors as pluripotent stem cell markers. These cells efficiently differentiated into myotubes after myogenic induction, at which point Nanog and Sox2 were down-regulated, whereas MyoD, myogenin, desmin and dystrophin were up-regulated. To our knowledge, this is the first demonstration that FTCVs can be efficiently directed to differentiate in vitro into skeletal muscle cells that express dystrophin as the last stage marker of myogenic differentiation. The myogenic potential of FTCVs reveals their promise for use in cell therapy for DMD, for which no effective treatment presently exists.
Collapse
Affiliation(s)
- Reiko Arakawa
- Affiliated Field of Medical Genetics, Division of Biomedical Engineering and Science, Graduate Course of Medicine, Graduate School of Tokyo Women's Medical University, Tokyo, Japan
| | | | | | | |
Collapse
|
27
|
Baron D, Magot A, Ramstein G, Steenman M, Fayet G, Chevalier C, Jourdon P, Houlgatte R, Savagner F, Pereon Y. Immune response and mitochondrial metabolism are commonly deregulated in DMD and aging skeletal muscle. PLoS One 2011; 6:e26952. [PMID: 22096509 PMCID: PMC3212519 DOI: 10.1371/journal.pone.0026952] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 10/06/2011] [Indexed: 01/12/2023] Open
Abstract
Duchenne Muscular Dystrophy (DMD) is a complex process involving multiple pathways downstream of the primary genetic insult leading to fatal muscle degeneration. Aging muscle is a multifactorial neuromuscular process characterized by impaired muscle regeneration leading to progressive atrophy. We hypothesized that these chronic atrophying situations may share specific myogenic adaptative responses at transcriptional level according to tissue remodeling. Muscle biopsies from four young DMD and four AGED subjects were referred to a group of seven muscle biopsies from young subjects without any neuromuscular disorder and explored through a dedicated expression microarray. We identified 528 differentially expressed genes (out of 2,745 analyzed), of which 328 could be validated by an exhaustive meta-analysis of public microarray datasets referring to DMD and Aging in skeletal muscle. Among the 328 validated co-expressed genes, 50% had the same expression profile in both groups and corresponded to immune/fibrosis responses and mitochondrial metabolism. Generalizing these observed meta-signatures with large compendia of public datasets reinforced our results as they could be also identified in other pathological processes and in diverse physiological conditions. Focusing on the common gene signatures in these two atrophying conditions, we observed enrichment in motifs for candidate transcription factors that may coordinate either the immune/fibrosis responses (ETS1, IRF1, NF1) or the mitochondrial metabolism (ESRRA). Deregulation in their expression could be responsible, at least in part, for the same transcriptome changes initiating the chronic muscle atrophy. This study suggests that distinct pathophysiological processes may share common gene responses and pathways related to specific transcription factors.
Collapse
|
28
|
White JD, Grounds MD. Harnessing the therapeutic potential of myogenic stem cells. Cytotechnology 2011; 41:153-64. [PMID: 19002952 DOI: 10.1023/a:1024830924103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The potential clinical use of stem cells for cell transplantation therapies to replace defective genes in myopathies is an area of intense investigation. Precursor cells derived from non-muscle tissue with myogenic potential have been identified in many tissues, including bone marrow and dermis, although the status of these putative stem cells requires clarification. The incorporation of circulating bone-marrow derived stem cells into regenerating adult skeletal muscle has been demonstrated in mice but the contribution of donor cells is so minimal that it would appear clinically irrelevant at this stage. The possibility of a true stem cell subpopulation within skeletal muscle that replenishes the satellite cells (conventional muscle precursors on the surface of myofibres) is also very attractive as a superior source of myoblasts for muscle construction. A full understanding of the intrinsic factors (i.e. gene expression within the stem cell) and extrinsic factors (i.e. signals from the external environment) which control the commitment of stem cells to the myogenic lineage, and the conditions which favour stem cell expansion in vivo is required before stem cells can be seriously considered for clinical cell therapy.
Collapse
Affiliation(s)
- Jason D White
- School of Anatomy and Human Biology, The University of Western Australia, Crawley, Perth, Western Australia, Australia (Author for correspondence; E-mail,
| | | |
Collapse
|
29
|
Cui CH, Miyoshi S, Tsuji H, Makino H, Kanzaki S, Kami D, Terai M, Suzuki H, Umezawa A. Dystrophin conferral using human endothelium expressing HLA-E in the non-immunosuppressive murine model of Duchenne muscular dystrophy. Hum Mol Genet 2011; 20:235-44. [PMID: 20947660 PMCID: PMC3005899 DOI: 10.1093/hmg/ddq458] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 09/16/2010] [Accepted: 10/08/2010] [Indexed: 12/17/2022] Open
Abstract
Human leukocyte antigen (HLA)-E is a non-classical major histocompatibility complex class I (Ib) molecule, which plays an important role in immunosuppression. In this study, we investigated the immunomodulating effect of HLA-E in a xenogeneic system, using human placental artery-derived endothelial (hPAE) cells expressing HLA-E in a mouse model. In vitro cell lysis analysis by primed lymphocytes in combination with siRNA transfection showed that HLA-E is necessary for inhibition of the immune response. Similarly, in vivo cell implantation analysis with siRNA-mediated down-regulation of HLA-E demonstrates that HLA-E is involved in immunosuppression. As hPAE cells efficiently transdifferentiate into myoblasts/myocytes in vitro, we transplanted the cells into mdx mice, a model of Duchenne muscular dystrophy. hPAE cells conferred dystrophin to myocytes of the 'immunocompetent' mdx mice with extremely high efficiency. These findings suggest that HLA-E-expressing cells with a myogenic potential represent a promising source for cell-based therapy of patients with muscular dystrophy.
Collapse
Affiliation(s)
- Chang-Hao Cui
- Department of Reproductive Biology, National Institute for Child Health and Development, Tokyo 157-8535, Japan,
- Department of Basic Medical Science, Mudanjiang Medical College, Mudanjiang 157011, China,
| | - Shunichiro Miyoshi
- Department of Cardiology, Keio University School of Medicine, Tokyo 160-8582, Japan and
| | - Hiroko Tsuji
- Department of Cardiology, Keio University School of Medicine, Tokyo 160-8582, Japan and
| | - Hatsune Makino
- Department of Reproductive Biology, National Institute for Child Health and Development, Tokyo 157-8535, Japan,
| | - Seiichi Kanzaki
- Department of Reproductive Biology, National Institute for Child Health and Development, Tokyo 157-8535, Japan,
| | - Daisuke Kami
- Department of Reproductive Biology, National Institute for Child Health and Development, Tokyo 157-8535, Japan,
| | - Masanori Terai
- Department of Reproductive Biology, National Institute for Child Health and Development, Tokyo 157-8535, Japan,
| | - Harumi Suzuki
- Department of Pathology, Research Institute, International Medical Center of Japan, Tokyo 162-8655, Japan
| | - Akihiro Umezawa
- Department of Reproductive Biology, National Institute for Child Health and Development, Tokyo 157-8535, Japan,
| |
Collapse
|
30
|
Meng J, Muntoni F, Morgan JE. Stem cells to treat muscular dystrophies – Where are we? Neuromuscul Disord 2011; 21:4-12. [DOI: 10.1016/j.nmd.2010.10.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 09/13/2010] [Accepted: 10/08/2010] [Indexed: 12/18/2022]
|
31
|
Liao IC, Leong KW. Efficacy of engineered FVIII-producing skeletal muscle enhanced by growth factor-releasing co-axial electrospun fibers. Biomaterials 2010; 32:1669-77. [PMID: 21084118 DOI: 10.1016/j.biomaterials.2010.10.049] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Accepted: 10/22/2010] [Indexed: 01/17/2023]
Abstract
Co-axial electrospun fibers can offer both topographical and biochemical cues for tissue engineering applications. In this study, we demonstrate the sustained treatment of hemophilia through a non-viral, tissue engineering approach facilitated by growth factor-releasing co-axial electrospun fibers. FVIII-producing skeletal myotubes were first engineered on aligned electrospun fibers in vitro, followed by implantation in hemophilic mice with or without a layer of core-shell electrospun fibers designed to provide sustained delivery of angiogenic or lymphangiogenic growth factors, which serves to stimulate the lymphatic or vascular systems to enhance the FVIII transport from the implant site into systemic circulation. Upon subcutaneous implantation into hemophilic mice, the construct seamlessly integrated with the host tissue within one month, and specifically induced either vascular or lymphatic network infiltration in accordance with the growth factors released from the electrospun fibers. Engineered constructs that induced angiogenesis resulted in sustained elevation of plasma FVIII and significantly reduced blood coagulation time for at least 2-months. Biomaterials-assisted functional tissue engineering was shown in this study to offer protein replacement therapy for a genetic disorder such as hemophilia.
Collapse
Affiliation(s)
- I-Chien Liao
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, Box 90281, Durham, NC 27708, USA.
| | | |
Collapse
|
32
|
Li Z, Liu HY, Lei QF, Zhang C, Li SN. Improved motor function in dko mice by intravenous transplantation of bone marrow-derived mesenchymal stromal cells. Cytotherapy 2010; 13:69-77. [PMID: 20735169 DOI: 10.3109/14653249.2010.510502] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS We explored the potential therapeutic value of transplanting bone marrow (BM)-derived mesenchymal stromal cells (MSC) into utrophin/dystrophin-deficient double knock-out (dko) mice, a murine model of Duchenne muscular dystrophy. METHODS MSC from male rats were isolated and transplanted into female dko mice via the caudal vein. Behavior and locomotor function were later evaluated, along with the expression of dystrophin and utrophin in the sarcolemma of myofiber tissues. The presence of grafted cells was confirmed via polymerase chain reaction for the sex-determining region of the Y-chromosome. RESULTS Locomotor activity improved significantly (P < 0.05) from 5 to 15 weeks after cell transplantation, as measured by traction, rotating rod and running wheel tests. We also found that the expression of dystrophin and utrophin increased significantly (P < 0.05) and progressively in the sarcolemma from 5 to 15 weeks after transplantation. The median lifespan of mice in the normal group (74.1 weeks) was significantly (P < 0.001) higher than those in the control (22.0 weeks) and transplantation (35.0 weeks) groups, and the median lifespan of mice in the transplantation group was significantly (P < 0.001) higher than that in the control group. CONCLUSIONS Results of this study demonstrate that BM MSC have potential value in xenogeneic transplantation therapy for muscular dystrophy.
Collapse
Affiliation(s)
- Zhong Li
- Department of Neurology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | | | | | | | | |
Collapse
|
33
|
Kawamichi Y, Cui CH, Toyoda M, Makino H, Horie A, Takahashi Y, Matsumoto K, Saito H, Ohta H, Saito K, Umezawa A. Cells of extraembryonic mesodermal origin confer human dystrophin in the mdx model of Duchenne muscular dystrophy. J Cell Physiol 2010; 223:695-702. [PMID: 20162617 DOI: 10.1002/jcp.22076] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Duchenne muscular dystrophy is an X-linked recessive genetic disease characterized by severe skeletal muscular degeneration. The placenta is considered to be a promising candidate cell source for cellular therapeutics because it contains a large number of cells and heterogenous cell populations with myogenic potentials. We analyzed the myogenic potential of cells obtained from six parts of the placenta, that is, umbilical cord, amniotic epithelium, amniotic mesoderm, chorionic plate, villous chorion, and decidua basalis. In vitro cells derived from amniotic mesoderm, chorionic plate, and villous chorion efficiently transdifferentiate into myotubes. In addition, in vivo implantation of placenta-derived cells into dystrophic muscles of immunodeficient mdx mice restored sarcolemmal expression of human dystrophin. Differential contribution to myogenesis in this study may be attributed to placental portion-dependent default cell state. Molecular taxonomic characterization of placenta-derived maternal and fetal cells in vitro will help determine the feasibility of cell-based therapy.
Collapse
Affiliation(s)
- Yayoi Kawamichi
- Department of Reproductive Biology, National Institute for Child Health and Development, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Ichim TE, Alexandrescu DT, Solano F, Lara F, Campion RDN, Paris E, Woods EJ, Murphy MP, Dasanu CA, Patel AN, Marleau AM, Leal A, Riordan NH. Mesenchymal stem cells as anti-inflammatories: implications for treatment of Duchenne muscular dystrophy. Cell Immunol 2010; 260:75-82. [PMID: 19917503 DOI: 10.1016/j.cellimm.2009.10.006] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Accepted: 10/13/2009] [Indexed: 01/01/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a lethal X-linked musculodegenerative condition consisting of an underlying genetic defect whose manifestation is augmented by inflammatory mechanisms. Previous treatment approaches using gene replacement, exon-skipping or allogeneic cell therapy have been relatively unsuccessful. The only intervention to mediate improvement in survival, albeit minor, is glucocorticoid treatment. Given this modality appears to function via suppression of underlying inflammation; we focus this review on the inflammatory response as a target for mesenchymal stem cell (MSC) therapy. In contrast to other cell based therapies attempted in DMD, MSC have the advantages of (a) ability to fuse with and genetically complement dystrophic muscle; (b) possess anti-inflammatory activities; and (c) produce trophic factors that may augment activity of endogenous repair cells. We conclude by describing one practical scenario of stem cell therapy for DMD.
Collapse
|
35
|
Bex1 Participates in Muscle Regeneration by Regulating Myogenic Satellite Cell Differentiation. Lab Anim Res 2010. [DOI: 10.5625/lar.2010.26.2.145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
36
|
Fan J, Varshney RR, Ren L, Cai D, Wang DA. Synovium-derived mesenchymal stem cells: a new cell source for musculoskeletal regeneration. TISSUE ENGINEERING PART B-REVIEWS 2009; 15:75-86. [PMID: 19196118 DOI: 10.1089/ten.teb.2008.0586] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Ever since synovium-derived mesenchymal stem cells (SMSCs) were first identified and successfully isolated in 2001, as a brand new member in MSC families, they have been increasingly regarded as a promising therapeutic cell species for musculoskeletal regeneration, particularly for reconstructions of cartilage, bones, tendons, and muscles. Besides the general multipotency in common among the MSC community, SMSCs excel other sourced MSCs in higher ability of proliferation and superiority in chondrogenesis. This review summarizes the latest advances in SMSC-related studies covering their specific isolation methodologies, biological insights, and practical applications in musculoskeletal therapeutics of which an emphasis is cast on engineered chondrogenesis.
Collapse
Affiliation(s)
- Jiabing Fan
- Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Guangzhou, PR China
| | | | | | | | | |
Collapse
|
37
|
Seidel M, Borczyńska A, Rozwadowska N, Kurpisz M. Cell-based therapy for heart failure: skeletal myoblasts. Cell Transplant 2009; 18:695-707. [PMID: 19500482 DOI: 10.3727/096368909x470810] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Satellite cells are committed precursor cells residing in the skeletal muscle. These cells provide an almost unlimited regeneration potential to the muscle, contrary to the heart, which, although proved to contain cardiac stem cells, possesses a very limited ability for self-renewal. The idea that myoblasts (satellite cell progenies) may repopulate postinfarction scar occurred around the mid-1990s. Encouraging results of preclinical studies triggered extensive research, which led to the onset of clinical trials. These trials have shown that autologous skeletal myoblast transplantation to cure heart failure is feasible and relatively safe (observed incidences of arrhythmia). Because most of the initial studies on myoblast application into postischemic heart have been carried out as an adjunct to routine surgical procedures, the true clinical outcome of such therapy in regard to cell implantation is blurred and requires to be elucidated. The mechanism by which implantation of skeletal myoblast may improve heart function is not clear, especially in the light of inability of these cells to couple electromechanically with a host myocardium. Successful myoblast therapy depends on a number of factors, including: delivery to the target tissue, long-term survival, efficacious engraftment, differentiation into cardiomyocytes, and integration into the new, unique microenvironment. All these steps constitute a potential goal for cell manipulation aiming to improve the overall outcome of such therapy. Precise understanding of the mechanism by which cells improve cardiac function is essential in giving the sensible direction of further research.
Collapse
Affiliation(s)
- Monika Seidel
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland
| | | | | | | |
Collapse
|
38
|
Tong JF, Yan X, Zhu MJ, Ford SP, Nathanielsz PW, Du M. Maternal obesity downregulates myogenesis and beta-catenin signaling in fetal skeletal muscle. Am J Physiol Endocrinol Metab 2009; 296:E917-24. [PMID: 19176350 PMCID: PMC2670630 DOI: 10.1152/ajpendo.90924.2008] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Skeletal muscle is one of the primary tissues responsible for insulin resistance and type 2 diabetes (T2D). The fetal stage is crucial for skeletal muscle development. Obesity induces inflammatory responses, which might regulate myogenesis through Wnt/beta-catenin signaling. This study evaluated the effects of maternal obesity (>30% increase in body mass index) during pregnancy on myogenesis and the Wnt/beta-catenin and IKK/NF-kappaB pathways in fetal skeletal muscle using an obese pregnant sheep model. Nonpregnant ewes were assigned to a control group (C; fed 100% of National Research Council recommendations; n=5) or obesogenic (OB; fed 150% of National Research Council recommendations; n=5) diet from 60 days before to 75 days after conception (term approximately 148 days) when fetal semitendenosus skeletal muscle was sampled for analyses. Myogenic markers including MyoD, myogenin, and desmin contents were reduced in OB compared with C fetal semitendenosus, indicating the downregulation of myogenesis. The diameter of primary muscle fibers was smaller in OB fetal muscle. Phosphorylation of GSK3beta was reduced in OB compared with C fetal semitendenosus. Although the beta-catenin level was lower in OB than C fetal muscle, more beta-catenin was associated with FOXO3a in the OB fetuses. Moreover, we found phosphorylation levels of IKKbeta and RelA/p65 were both increased in OB fetal muscle. In conclusion, our data showed that myogenesis and the Wnt/beta-catenin signaling pathway were downregulated, which might be due to the upregulation of inflammatory IKK/NF-kappaB signaling pathways in fetal muscle of obese mothers.
Collapse
Affiliation(s)
- Jun F Tong
- Center for the Study of Fetal Programming, Department of Animal Science, University of Wyoming, Laramie, WY 82071, USA
| | | | | | | | | | | |
Collapse
|
39
|
Direct muscle delivery of GDNF with human mesenchymal stem cells improves motor neuron survival and function in a rat model of familial ALS. Mol Ther 2008; 16:2002-10. [PMID: 18797452 DOI: 10.1038/mt.2008.197] [Citation(s) in RCA: 197] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease in which there is a progressive loss of motor neurons and their connections to muscle, leading to paralysis. In order to maintain muscle connections in a rat model of familial ALS (FALS), we performed intramuscular transplantation with human mesenchymal stem cells (hMSCs) used as "Trojan horses" to deliver growth factors to the terminals of motor neurons and to the skeletal muscles. hMSCs engineered to secrete glial cell line-derived neurotrophic factor (hMSC-GDNF) were transplanted bilaterally into three muscle groups. The cells survived within the muscle, released GDNF, and significantly increased the number of neuromuscular connections and motor neuron cell bodies in the spinal cord at mid-stages of the disease. Further, intramuscular transplantation with hMSC-GDNF was found to ameliorate motor neuron loss within the spinal cord where it connects with the limb muscles receiving transplants. While disease onset was similar in all the animals, hMSC-GDNF significantly delayed disease progression, increasing overall lifespan by up to 28 days, which is one of the largest effects on survival noted for this rat model of FALS. This preclinical data provides a novel and practical approach toward ex vivo gene therapy for ALS.
Collapse
|
40
|
Straface G, Aprahamian T, Flex A, Gaetani E, Biscetti F, Smith RC, Pecorini G, Pola E, Angelini F, Stigliano E, Castellot JJ, Losordo DW, Pola R. Sonic hedgehog regulates angiogenesis and myogenesis during post-natal skeletal muscle regeneration. J Cell Mol Med 2008; 13:2424-2435. [PMID: 18662193 DOI: 10.1111/j.1582-4934.2008.00440.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Sonic hedgehog (Shh) is a morphogen-regulating crucial epithelial-mesenchymal interactions during embryonic development, but its signalling pathway is considered generally silent in post-natal life. In this study, we demonstrate that Shh is de novo expressed after injury and during regeneration of the adult skeletal muscle. Shh expression is followed by significant up-regulation of its receptor and target gene Ptc1 in injured and regenerating muscles. The reactivation of the Shh signalling pathway has an important regulatory role on injury-induced angiogenesis, as inhibition of Shh function results in impaired up-regulation of prototypical angiogenic agents, such as vascular endothelial growth factor (VEGF) and stromal-derived factor (SDF)-1alpha, decreased muscle blood flow and reduced capillary density after injury. In addition, Shh reactivation plays a regulatory role on myogenesis, as its inhibition impairs the activation of the myogenic regulatory factors Myf-5 and MyoD, decreases the up-regulation of insulin-like growth factor (IGF)-1 and reduces the number of myogenic satellite cells at injured site. Finally, Shh inhibition results in muscle fibrosis, increased inflammatory reaction and compromised motor functional recovery after injury. These data demonstrate that the Shh pathway is functionally important for adult skeletal muscle regeneration and displays pleiotropic angiogenic and myogenic potentials in post-natal life. These findings might constitute the foundation for new therapeutic approaches for muscular diseases in humans.
Collapse
Affiliation(s)
- Giuseppe Straface
- Laboratory of Vascular Biology and Genetics, Department of Medicine, Catholic University School of Medicine, Rome, Italy
| | - Tamar Aprahamian
- Renal Section, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | - Andrea Flex
- Laboratory of Vascular Biology and Genetics, Department of Medicine, Catholic University School of Medicine, Rome, Italy
| | - Eleonora Gaetani
- Laboratory of Vascular Biology and Genetics, Department of Medicine, Catholic University School of Medicine, Rome, Italy
| | - Federico Biscetti
- Laboratory of Vascular Biology and Genetics, Department of Medicine, Catholic University School of Medicine, Rome, Italy
| | - Roy C Smith
- Center of Cardiovascular Research, Department of Medicine, Caritas St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Giovanni Pecorini
- Laboratory of Vascular Biology and Genetics, Department of Medicine, Catholic University School of Medicine, Rome, Italy
| | - Enrico Pola
- Department of Orthopedics, Catholic University School of Medicine, Rome, Italy
| | - Flavia Angelini
- Laboratory of Vascular Biology and Genetics, Department of Medicine, Catholic University School of Medicine, Rome, Italy
| | - Egidio Stigliano
- Department of Pathology, Catholic University School of Medicine, Rome, Italy
| | - John J Castellot
- Department of Anatomy and Cell Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Douglas W Losordo
- Feinberg Cardiovascular Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| | - Roberto Pola
- Laboratory of Vascular Biology and Genetics, Department of Medicine, Catholic University School of Medicine, Rome, Italy.,Center of Cardiovascular Research, Department of Medicine, Caritas St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA, USA.,IRCCS OASI, Troina, Italy
| |
Collapse
|
41
|
Lorenzi B, Pessina F, Lorenzoni P, Urbani S, Vernillo R, Sgaragli G, Gerli R, Mazzanti B, Bosi A, Saccardi R, Lorenzi M. Treatment of experimental injury of anal sphincters with primary surgical repair and injection of bone marrow-derived mesenchymal stem cells. Dis Colon Rectum 2008; 51:411-20. [PMID: 18224375 DOI: 10.1007/s10350-007-9153-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2007] [Revised: 07/16/2007] [Accepted: 08/25/2007] [Indexed: 12/15/2022]
Abstract
PURPOSE Sphincter injury is a common cause of anal incontinence. Surgical repair remains the operation of choice; however, the outcome often is poor. We investigated the ability of injected bone marrow-derived mesenchymal stem cells to enhance sphincter healing after injury and primary repair in a preclinical model. METHODS Twenty-four inbred Wistar Furth rats were divided into three groups. As a control, Group A underwent sham operation. Group B had sphincterotomy and repair of both anal sphincters plus saline injections. The study group (Group C) underwent sphincterotomy and repair followed by intrasphincteric injections of syngenic bone marrow-derived mesenchymal stem cells. A further group (Group D) of outbred Wistar rats treated with mesenchymal stem cells and immunosuppressive therapy also was evaluated. At 30 days, histologic and morphometric analysis and in vitro contractility testing was performed. RESULTS A significant decrease of muscle tissue was observed at the site of repair after sphincter injury. However, in Groups C and D, histologic examination demonstrated new muscle fibers and morphometric analysis revealed a significantly greater muscle area fraction than in Group B (P < 0.05). Moreover, mesenchymal stem cells injection improved contractility of sphincters strips compared with Group B (P < 0.05). No significant differences were found between Groups C and D. CONCLUSIONS In our experimental model, bone marrow-derived mesenchymal stem cells injection improved muscle regeneration and increased contractile function of anal sphincters after injury and repair. Therefore, mesenchymal stem cells may represent an attractive tool for treating anal sphincter lesions in humans. Investigations into the biologic basis of this phenomenon should increase our knowledge on underlying mechanisms involved in sphincter repair.
Collapse
Affiliation(s)
- Bruno Lorenzi
- Department of Surgery, University of Siena, Viale Bracci, 53100 Siena, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Liu Y, Yan X, Sun Z, Chen B, Han Q, Li J, Zhao RC. Flk-1+ adipose-derived mesenchymal stem cells differentiate into skeletal muscle satellite cells and ameliorate muscular dystrophy in mdx mice. Stem Cells Dev 2008; 16:695-706. [PMID: 17999592 DOI: 10.1089/scd.2006.0118] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe hereditary disease characterized by the absence of dystrophin on the sarcolemma of muscle fiber. This absence results in widespread muscle damage and satellite cell activation. After depletion of the satellite cell pool, skeletal muscle is then invariably replaced by connective tissue, leading to progressive muscle weakness. Herein, we isolated Flk-1(+) mesenchymal stem cells (MSCs) from adult adipose tissue and induced them to differentiate into skeletal muscle cells in culture. Within mdx mice, an animal model of DMD, adipose tissue-derived Flk-1(+) MSCs (AD-MSCs) homed to and differentiated into cells that repaired injured muscle tissue. This repair correlated with reconstitution of dystrophin expression on the damaged fibers. Flk-1(+) AD-MSCs also differentiated into muscle satellite cells. This differentiation may have accounted for long-term reconstitution. These cells also differentiated into endothelial cells, thereby possibly improving fiber regeneration as a result of the induced angiogenesis. Therefore, Flk-1(+) AD-MSC transplants may repair muscular dystrophy.
Collapse
Affiliation(s)
- Yanning Liu
- Institute of Basic Medical Sciences & School of Basic Medicine, Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | | | | | | | | | | | | |
Collapse
|
43
|
Shang YC, Zhang C, Wang SH, Xiong F, Zhao CP, Peng FN, Feng SW, Yu MJ, Li MS, Zhang YN, Li Y. Activated beta-catenin induces myogenesis and inhibits adipogenesis in BM-derived mesenchymal stromal cells. Cytotherapy 2008; 9:667-81. [PMID: 17917885 DOI: 10.1080/14653240701508437] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Mesenchymal stromal cells (MSC) have been thought to be attractive candidates for the treatment of degenerative muscle diseases. However, little is known about the molecular mechanisms governing the myogenic differentiation in MSC. As the Wnt signaling pathway has been associated with myogenesis in embryogenesis and post-natal muscle regeneration, we hypothesized that the Wnt signaling pathway may be involved in governing the myogenic differentiation in MSC. METHODS Primary MSC were isolated from Sprague-Dawley rats and expanded in proliferation medium. The rMSC were transfected with a constitutively active hbeta-catenin (S37A) plasmid or control vector by Lipofectamine followed by G418 selection. The transfected rMSC were grown to 80% confluence and then cultured in myogenic or adipogenic differentiation medium. Cells were characterized by light microscopy, immunofluorescence and RT-PCR at different time points after myogenic or adipogenic introduction. RESULTS Ectopic expression of activated beta-catenin located primarily in the nucleus and activated transcription in rMSC. Overexpression of stabilized beta-catenin induced 27.1 +/- 3.91% rMSC forming long multinucleated cells expressing MyoD, myogenin, desmin and myosin heavy chain (MHC) via evoking the expression of skeletal muscle-specific transcription factors. In addition, overexpression of activated beta-catenin inhibited the adipogenic differentiation in rMSC through down-regulated expressions of C/EBPalpha and PPARgamma. DISCUSSION To our knowledge, this is the first evidence that activated beta-catenin can induce myogenic differentiation in rMSC. The ability of stabilized beta-catenin to induce myogenic differentiation in rMSC may allow for its therapeutic application.
Collapse
Affiliation(s)
- Y C Shang
- Department of Neurology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Cossu G, Sampaolesi M. New therapies for Duchenne muscular dystrophy: challenges, prospects and clinical trials. Trends Mol Med 2007; 13:520-6. [PMID: 17983835 DOI: 10.1016/j.molmed.2007.10.003] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2007] [Revised: 10/10/2007] [Accepted: 10/10/2007] [Indexed: 02/05/2023]
Abstract
Muscular dystrophies primarily affect skeletal muscle. Mutations in a large number of genes, mainly encoding cytoskeletal proteins, cause different forms of dystrophy that compromise patient mobility and quality of life, and in the most severe cases lead to complete paralysis and premature death. Although muscular dystrophies still lack an effective therapy, several novel strategies are entering or are ready to enter clinical trials. Here we review the main experimental strategies, namely drug, gene and cell therapies, outlining their goals and limitations. We also provide an update of ongoing or planned clinical trials based on these strategies.
Collapse
Affiliation(s)
- Giulio Cossu
- Stem Cell Research Institute, Dibit, H. San Raffaele, 58 Via Olgettina, 20132 Milan, Italy.
| | | |
Collapse
|
45
|
Thibaud JL, Monnet A, Bertoldi D, Barthélémy I, Blot S, Carlier PG. Characterization of dystrophic muscle in golden retriever muscular dystrophy dogs by nuclear magnetic resonance imaging. Neuromuscul Disord 2007; 17:575-84. [PMID: 17537632 DOI: 10.1016/j.nmd.2007.03.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2006] [Revised: 03/08/2007] [Accepted: 03/27/2007] [Indexed: 10/23/2022]
Abstract
The Golden Retriever Muscular Dystrophy dog lacks dystrophin. Disease progression in this model shares many similarities with the Duchenne muscular dystrophy, both from anatomico pathological and clinical standpoints. The model is increasingly used in pre-clinical trials but needs to be further investigated, particularly with reference to the evaluation of therapies. The aim of this study was to identify quantitative indices that would help characterize the dystrophic dog non-invasively using NMR imaging. Two-month-old dystrophic dogs and healthy control animals were scanned at 4T. Standard T2- and T1-weighted images, fat-saturated T1-weighted images pre- and post-gadolinium chelate injection were acquired and kinetics of muscle enhancement were studied over a 2-h period. Several indices were found to be abnormally high in dystrophic dogs: the T2-weighted/T1-weighted signal ratio, T2-weighted image heterogeneity and maximal signal enhancement post-gadolinium. These may be proposed to evaluate muscle structural alterations non-invasively in this disease.
Collapse
Affiliation(s)
- J-L Thibaud
- Neurobiology Laboratory, National Veterinary School of Alfort, Maisons-Alfort, France and INSERM EMI 00-11, Créteil, France
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
The neuromuscular disorders are associated with diminished cardiopulmonary reserves, deficient airway protection mechanisms, and atypical responses to drugs used during anesthesia. Many of these conditions are uncommon, and methodologically sound evidence to guide clinical practice is limited. The disorders discussed in the present review are the motor neuron diseases, peripheral neuropathies, myasthenic syndromes, and myopathies, including malignant hyperthermia. Recent data on pathogenesis and medical management are outlined, as are studies relating to anesthesia and the perioperative period.
Collapse
Affiliation(s)
- R D Stevens
- Department of Anesthesia and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland 21287, USA.
| |
Collapse
|
47
|
Boldrin L, Elvassore N, Malerba A, Flaibani M, Cimetta E, Piccoli M, Baroni MD, Gazzola MV, Messina C, Gamba P, Vitiello L, De Coppi P. Satellite cells delivered by micro-patterned scaffolds: a new strategy for cell transplantation in muscle diseases. ACTA ACUST UNITED AC 2007; 13:253-62. [PMID: 17504060 DOI: 10.1089/ten.2006.0093] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Myoblast transplantation is a potentially useful therapeutic tool in muscle diseases, but the lack of an efficient delivery system has hampered its application. Here we have combined cell biology and polymer processing to create an appropriate microenvironment for in vivo transplantation of murine satellite cells (mSCs). Cells were prepared from single muscle fibers derived from C57BL/6-Tgn enhanced green fluorescent protein (GFP) transgenic mice. mSCs were expanded and seeded within micro-patterned polyglycolic acid 3-dimensional scaffolds fabricated using soft lithography and thermal membrane lamination. Myogenicity was then evaluated in vitro using immunostaining, flow cytometry, and reverse transcription polymerase chain reaction analyses. Scaffolds containing mSCs were implanted in pre-damaged tibialis anterior muscles of GFP-negative syngenic mice. Cells detached from culture dishes were directly injected into contra-lateral limbs as controls. In both cases, delivered cells participated in muscle regeneration, although scaffold-implanted muscles showed a much higher number of GFP-positive fibers in CD57 mice. These findings suggest that implantation of cellularized scaffolds is better than direct injection for delivering myogenic cells into regenerating skeletal muscle.
Collapse
Affiliation(s)
- Luisa Boldrin
- Stem Cell Processing Laboratory, Department of Pediatrics, University of Padova, Padova, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Wong SHA, Lowes KN, Bertoncello I, Quigley AF, Simmons PJ, Cook MJ, Kornberg AJ, Kapsa RMI. Evaluation of Sca-1 and c-Kit As Selective Markers for Muscle Remodelling by Nonhemopoietic Bone Marrow Cells. Stem Cells 2007; 25:1364-74. [PMID: 17303817 DOI: 10.1634/stemcells.2006-0194] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Bone marrow (BM)-derived cells (BMCs) have demonstrated a myogenic tissue remodeling capacity. However, because the myoremodeling is limited to approximately 1%-3% of recipient muscle fibers in vivo, there is disagreement regarding the clinical relevance of BM for therapeutic application in myodegenerative conditions. This study sought to determine whether rare selectable cell surface markers (in particular, c-Kit) could be used to identify a BMC population with enhanced myoremodeling capacity. Dystrophic mdx muscle remodeling has been achieved using BMCs sorted by expression of stem cell antigen-1 (Sca-1). The inference that Sca-1 is also a selectable marker associated with myoremodeling capacity by muscle-derived cells prompted this study of relative myoremodeling contributions from BMCs (compared with muscle cells) on the basis of expression or absence of Sca-1. We show that myoremodeling activity does not differ in cells sorted solely on the basis of Sca-1 from either muscle or BM. In addition, further fractionation of BM to a more mesenchymal-like cell population with lineage markers and CD45 subsequently revealed a stronger selectability of myoremodeling capacity with c-Kit/Sca-1 (p < .005) than with Sca-1 alone. These results suggest that c-Kit may provide a useful selectable marker that facilitates selection of cells with an augmented myoremodeling capacity derived from BM and possibly from other nonmuscle tissues. In turn, this may provide a new methodology for rapid isolation of myoremodeling capacities from muscle and nonmuscle tissues. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Sharon H A Wong
- National Muscular Dystrophy Research Centre, Department of Clinical Neurosciences, St. Vincent's Hospital, 35 Victoria Parade, Fitzroy, Victoria, 3065, Australia
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Yu M, Zhang C, Zhang Y, Feng S, Yao X, Lu X. BM stem cell transplantation rescues pathophysiologic features of aged dystrophic mdx muscle. Cytotherapy 2007; 9:44-52. [PMID: 17354101 DOI: 10.1080/14653240601114815] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND The value of transplantation of BM stem cells in aged (12-month-old) mdx was evaluated because it is thought to be a more ideal model for studying the praxiology of Duchenne muscular dystrophy (DMD). The possible mechanisms of stem cell differentiation were then discussed. METHODS BM was isolated from 8-10-week-old male C57 BL/10 mice. After injecting BM cells into 12-month-old female mdx mice through the tail vein, the expression of dystrophin and MyoD was detected at different time points by immunofluorescence staining, RT-PCR and Western blot. RESULTS The C57 male mice donor-specific and Y-chromosome-specific sequence could be detected in all female aged mdx mice, implying the success of the transplantation. Expression of dystrophin and MyoD was detected and increased over time. DISCUSSION BM cells were recruited to the muscle and partially restored specific pathophysiologic features of the dystrophic muscle in aged mdx mice. Muscle differentiation of BM cells recapitulated embryonic myogenesis.
Collapse
Affiliation(s)
- M Yu
- Department of Neurology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
50
|
Martin CM, Russell JL, Ferdous A, Garry DJ. Molecular signatures define myogenic stem cell populations. ACTA ACUST UNITED AC 2007; 2:37-42. [PMID: 17142885 DOI: 10.1007/s12015-006-0007-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 10/23/2022]
Abstract
Developmental and regenerative mechanisms are directed by stem cell populations. Skeletal muscle is a dynamic tissue that is capable of adapting to stress and severe injury due to a resident somatic stem cell population. In response to a severe injury that destroys upward of 90% of the tissue, skeletal muscle efficiently and reproducibly regenerates damaged tissue and restores the cellular architecture within a 2-wk period. Recent studies have localized and examined the molecular regulation of skeletal muscle stem cell populations using emerging molecular biological technologies. These studies enhance the understanding of the regulatory mechanisms that direct the somatic stem cell populations and the role they play in development and regeneration. Furthermore, these basic science studies will serve as a platform for future therapies directed toward patients with myopathic diseases.
Collapse
Affiliation(s)
- Cindy M Martin
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | |
Collapse
|